1
|
Radisch S, Poltorak MP, Wagner M, Cletiu V, Radisch C, Treise I, Pann S, Weigt A, Artner S, Dreher S, Fechner F, Borjan B, Fraessle SP, Effenberger M, Benke E, Navratil G, Hentschel N, Busch DH, Schmidt T, Stemberger C, Germeroth L. Next generation automated traceless cell chromatography platform for GMP-compliant cell isolation and activation. Sci Rep 2022; 12:6572. [PMID: 35449227 PMCID: PMC9023455 DOI: 10.1038/s41598-022-10320-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
Large-scale target cell isolation from patient blood preparations is one of the critical operations during drug product manufacturing for personalized cell therapy in immuno-oncology. Use of high-affinity murine antibody coated magnetic nanoparticles that remain on isolated cells is the current standard applied for this purpose. Here, we present the transformation of previously described technology - non-magnetic immunoaffinity column chromatography-based cell selection with reversible reagents into a new clinical-grade cell isolation platform called Automated Traceless Cell affinity chromatography (ATC). ATC is a fully closed and GMP-compliant cell selection and manufacturing system. Reversibility of reagents enables (sequential) positive cell selection, optionally in combination with depletion columns, enabling capture of highly specific cell subsets. Moreover, synergy with other Streptamer-based technologies allows novel uses beyond cell isolation including integrated and automated on-column target cell activation. In conclusion, ATC technology is an innovative as well as versatile platform to select, stimulate and modify cells for clinical manufacturing and downstream therapies.
Collapse
Affiliation(s)
- Sabine Radisch
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Mateusz P Poltorak
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany.
| | - Michaela Wagner
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Vlad Cletiu
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Christian Radisch
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Irina Treise
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Steffi Pann
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Alexis Weigt
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Sophie Artner
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Stefan Dreher
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Fabian Fechner
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Bojana Borjan
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Simon P Fraessle
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Manuel Effenberger
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Eileen Benke
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Gottfried Navratil
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Norbert Hentschel
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Thomas Schmidt
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Christian Stemberger
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| | - Lothar Germeroth
- Juno Therapeutics GmbH, Bristol-Myers Squibb Company, Grillparzerstr. 10, 81675, Munich, Germany
| |
Collapse
|
2
|
Cryo-EM structures of intermediates suggest an alternative catalytic reaction cycle for cytochrome c oxidase. Nat Commun 2021; 12:6903. [PMID: 34824221 PMCID: PMC8617209 DOI: 10.1038/s41467-021-27174-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022] Open
Abstract
Cytochrome c oxidases are among the most important and fundamental enzymes of life. Integrated into membranes they use four electrons from cytochrome c molecules to reduce molecular oxygen (dioxygen) to water. Their catalytic cycle has been considered to start with the oxidized form. Subsequent electron transfers lead to the E-state, the R-state (which binds oxygen), the P-state (with an already split dioxygen bond), the F-state and the O-state again. Here, we determined structures of up to 1.9 Å resolution of these intermediates by single particle cryo-EM. Our results suggest that in the O-state the active site contains a peroxide dianion and in the P-state possibly an intact dioxygen molecule, the F-state may contain a superoxide anion. Thus, the enzyme's catalytic cycle may have to be turned by 180 degrees.
Collapse
|
3
|
Fields C, Li P, O'Mahony JJ, Lee GU. Advances in affinity ligand-functionalized nanomaterials for biomagnetic separation. Biotechnol Bioeng 2015; 113:11-25. [DOI: 10.1002/bit.25665] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/21/2015] [Accepted: 05/24/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Conor Fields
- School of Chemistry and Chemical Biology; University College Dublin, Belfield, Dublin 4; Ireland
| | - Peng Li
- School of Chemistry and Chemical Biology; University College Dublin, Belfield, Dublin 4; Ireland
| | - James J. O'Mahony
- School of Chemistry and Chemical Biology; University College Dublin, Belfield, Dublin 4; Ireland
| | - Gil U. Lee
- School of Chemistry and Chemical Biology; University College Dublin, Belfield, Dublin 4; Ireland
- Conway Institute for Biomolecular and Biomedical Sciences; University College Dublin, Belfield, Dublin 4; Ireland
| |
Collapse
|
4
|
Lu NN, Xie M, Wang J, Lv SW, Yi JS, Dong WG, Huang WH. Biotin-triggered decomposable immunomagnetic beads for capture and release of circulating tumor cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:8817-26. [PMID: 25853336 DOI: 10.1021/acsami.5b01397] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Isolation of rare, pure, and viable circulating tumor cells (CTCs) provides a significant insight in early cancer diagnosis, and release of captured CTCs without damage for ex vivo culture may offer an opportunity for personalized cancer therapy. In this work, we described a biotin-triggered decomposable immunomagnetic system, in which peptide-tagged antibody designed by chemical conjugation was specifically immobilized on engineered protein-coated magnetic beads. The interaction between peptide and engineered protein can be reversibly destroyed by biotin treatment, making capture and release of CTCs possible. Furthermore, the peptide could mediate multiple antibodies' coimmobilization on engineered protein-coated magnetic beads, by which capture efficiency for CTCs was obviously improved. Quantitative results showed that 70% of captured cells could be released by biotin addition, and 85% of released cells remained viable. In addition, 79% of cancer cells spiked in human whole blood were captured and could also be successfully released for culture. Finally, immunomagnetic beads simultaneously loaded with anti-EpCAM, anti-HER2, and anti-EGFR were successfully applied to isolate and detect CTCs in 17 cancer patients' peripheral blood samples, and 2-215 CTCs were identified with high purity. These results suggest that our method is reliable and has great potential in CTC detection for CTC-based molecular profiling, diagnosis, and therapy.
Collapse
Affiliation(s)
- Ning-Ning Lu
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Min Xie
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jing Wang
- ‡Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Song-Wei Lv
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jia-Sheng Yi
- ‡Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Guo Dong
- ‡Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei-Hua Huang
- †Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
5
|
Kohlstaedt M, von der Hocht I, Hilbers F, Thielmann Y, Michel H. Development of a Thermofluor assay for stability determination of membrane proteins using the Na(+)/H(+) antiporter NhaA and cytochrome c oxidase. ACTA ACUST UNITED AC 2015; 71:1112-22. [PMID: 25945577 DOI: 10.1107/s1399004715004058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/26/2015] [Indexed: 11/10/2022]
Abstract
Crystallization of membrane proteins is very laborious and time-consuming, yielding well diffracting crystals in only a minority of projects. Therefore, a rapid and easy method is required to optimize the conditions for initial crystallization trials. The Thermofluor assay has been developed as such a tool. However, its applicability to membrane proteins is still limited because either large hydrophilic extramembranous regions or cysteine residues are required for the available dyes to bind and therefore act as reporters in this assay. No probe has been characterized to discriminate between the hydrophobic surfaces of detergent micelles, folded and detergent-covered membrane proteins and denatured membrane proteins. Of the four dyes tested, the two dyes 1-anilinonaphthalene-8-sulfonic acid (ANS) and SYPRO Orange were systematically screened for compatibility with five detergents commonly used in the crystallization of membrane proteins. ANS showed the weakest interactions with all of the detergents screened. It was possible to determine the melting temperature of the sodium ion/proton antiporter NhaA, a small membrane protein without large hydrophilic domains, over a broad pH range using ANS. Furthermore, cytochrome c oxidase (CcO) was used to apply the method to a four-subunit membrane protein complex. It was possible to obtain preliminary information on the temperature-dependent denaturation of this complex using the dye ANS. Application of the dye 7-diethylamino-3-(4'-maleimidylphenyl)-4-methylcoumarin (CPM) to CcO in the Thermofluor assay enabled the determination of the melting temperatures of distinct subunits of the complex.
Collapse
Affiliation(s)
- Martin Kohlstaedt
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60438 Frankfurt, Germany
| | - Iris von der Hocht
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60438 Frankfurt, Germany
| | - Florian Hilbers
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60438 Frankfurt, Germany
| | - Yvonne Thielmann
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60438 Frankfurt, Germany
| | - Hartmut Michel
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60438 Frankfurt, Germany
| |
Collapse
|
6
|
Mir SH, Escher C, Kao WC, Birth D, Wirth C, Hunte C. Generation of recombinant antibody fragments for membrane protein crystallization. Methods Enzymol 2015; 557:201-18. [PMID: 25950966 DOI: 10.1016/bs.mie.2014.12.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Membrane proteins are challenging targets for crystallization and structure determination by X-ray crystallography. Hurdles can be overcome by antibody-mediated crystallization. More than 25 unique structures of membrane protein:antibody complexes have already been determined. In the majority of cases, hybridoma-derived antibody fragments either in Fab or Fv fragment format were employed for these complexes. We will briefly introduce the background and current status of the strategy and describe in detail the current protocols of well-established methods for the immunization, the selection, and the characterization of antibodies, as well as the cloning, the production, and the purification of recombinant antibodies useful for structural analysis of membrane proteins.
Collapse
Affiliation(s)
- Syed H Mir
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany; Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | - Claudia Escher
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Wei-Chun Kao
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Dominic Birth
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Christophe Wirth
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Carola Hunte
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
7
|
Electrochemistry suggests proton access from the exit site to the binuclear center in Paracoccus denitrificans cytochrome c oxidase pathway variants. FEBS Lett 2015; 589:565-8. [PMID: 25637325 DOI: 10.1016/j.febslet.2015.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 11/27/2022]
Abstract
Two different pathways through which protons access cytochrome c oxidase operate during oxygen reduction from the mitochondrial matrix, or the bacterial cytoplasm. Here, we use electrocatalytic current measurements to follow oxygen reduction coupled to proton uptake in cytochrome c oxidase isolated from Paracoccus denitrificans. Wild type enzyme and site-specific variants with defects in both proton uptake pathways (K354M, D124N and K354M/D124N) were immobilized on gold nanoparticles, and oxygen reduction was probed electrochemically in the presence of varying concentrations of Zn(2+) ions, which are known to inhibit both the entry and the exit proton pathways in the enzyme. Our data suggest that under these conditions substrate protons gain access to the oxygen reduction site via the exit pathway.
Collapse
|
8
|
Kozuch J, von der Hocht I, Hilbers F, Michel H, Weidinger IM. Resonance Raman characterization of the ammonia-generated oxo intermediate of cytochrome c oxidase from Paracoccus denitrificans. Biochemistry 2013; 52:6197-202. [PMID: 23914722 DOI: 10.1021/bi400535m] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A novel oxo state of cytochrome c oxidase from Paracoccus denitrificans generated by successive addition of excess H2O2 and ammonia was investigated using resonance Raman (RR) spectroscopy. Addition of ammonia to the H2O2-generated artificial F state resulted in an upshift of the oxoferryl stretching vibration from 790 to 796 cm(-1), indicating that ammonia influences ligation of the heme-bound oxygen in the binuclear center. Concomitantly performed RR measurements in the high-frequency region between 1300 and 1700 cm(-1) showed a high-spin to low-spin transition of heme a3 upon generation of the F state that was not altered by addition of ammonia. Removal of H2O2 by addition of catalase resulted in the disappearance of the oxoferryl stretching vibration and major back transformation of heme a3 into the high-spin state. The ratio of high-spin to low-spin states was identical for intermediates created with and without ammonia, leading to the conclusion that ammonia does not interact directly with heme a3. Only for the ammonia-created state was a band at 612 nm observed in the UV-visible difference spectrum that was shifted to 608 nm after addition of catalase. Our results support the hypothesis by von der Hocht et al. [von der Hocht, I., et al. (2011) Proc. Natl. Acad. Sci. U.S.A. 108, 3964-3969] that addition of ammonia creates a novel oxo intermediate state called PN where ammonia binds to CuB once the oxo intermediate F state has been formed.
Collapse
Affiliation(s)
- Jacek Kozuch
- Technische Universität Berlin , Institut fuer Chemie, Strasse des 17. Juni 135, 10623 Berlin, Germany
| | | | | | | | | |
Collapse
|
9
|
Kirchberg K, Michel H, Alexiev U. Exploring the entrance of proton pathways in cytochrome c oxidase from Paracoccus denitrificans: Surface charge, buffer capacity and redox-dependent polarity changes at the internal surface. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1827:276-84. [DOI: 10.1016/j.bbabio.2012.10.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/18/2012] [Accepted: 10/25/2012] [Indexed: 11/25/2022]
|
10
|
Kirchberg K, Michel H, Alexiev U. Net proton uptake is preceded by multiple proton transfer steps upon electron injection into cytochrome c oxidase. J Biol Chem 2012; 287:8187-93. [PMID: 22238345 DOI: 10.1074/jbc.m111.338491] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytochrome c oxidase (COX), the last enzyme of the respiratory chain of aerobic organisms, catalyzes the reduction of molecular oxygen to water. It is a redox-linked proton pump, whose mechanism of proton pumping has been controversially discussed, and the coupling of proton and electron transfer is still not understood. Here, we investigated the kinetics of proton transfer reactions following the injection of a single electron into the fully oxidized enzyme and its transfer to the hemes using time-resolved absorption spectroscopy and pH indicator dyes. By comparison of proton uptake and release kinetics observed for solubilized COX and COX-containing liposomes, we conclude that the 1-μs electron injection into Cu(A), close to the positive membrane side (P-side) of the enzyme, already results in proton uptake from both the P-side and the N (negative)-side (1.5 H(+)/COX and 1 H(+)/COX, respectively). The subsequent 10-μs transfer of the electron to heme a is accompanied by the release of 1 proton from the P-side to the aqueous bulk phase, leaving ∼0.5 H(+)/COX at this side to electrostatically compensate the charge of the electron. With ∼200 μs, all but 0.4 H(+) at the N-side are released to the bulk phase, and the remaining proton is transferred toward the hemes to a so-called "pump site." Thus, this proton may already be taken up by the enzyme as early as during the first electron transfer to Cu(A). These results support the idea of a proton-collecting antenna, switched on by electron injection.
Collapse
|
11
|
Lieberman RL, Culver JA, Entzminger KC, Pai JC, Maynard JA. Crystallization chaperone strategies for membrane proteins. Methods 2011; 55:293-302. [PMID: 21854852 DOI: 10.1016/j.ymeth.2011.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 08/03/2011] [Accepted: 08/05/2011] [Indexed: 10/17/2022] Open
Abstract
From G protein-coupled receptors to ion channels, membrane proteins represent over half of known drug targets. Yet, structure-based drug discovery is hampered by the dearth of available three-dimensional models for this large category of proteins. Other than efforts to improve membrane protein expression and stability, current strategies to improve the ability of membrane proteins to crystallize involve examining many orthologs and DNA constructs, testing the effects of different detergents for purification and crystallization, creating a lipidic environment during crystallization, and cocrystallizing with covalent or non-covalent soluble protein chaperones with an intrinsic high propensity to crystallize. In this review, we focus on this last category, highlighting successes of crystallization chaperones in membrane protein structure determination and recent developments in crystal chaperone engineering, including molecular display to enhance chaperone crystallizability, and end with a novel generic approach in development to target any membrane protein of interest.
Collapse
Affiliation(s)
- Raquel L Lieberman
- School of Chemistry and Biochemistry, Institute for Bioscience and Bioengineering, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | | | | | | | | |
Collapse
|
12
|
Interconversions of P and F intermediates of cytochrome c oxidase from Paracoccus denitrificans. Proc Natl Acad Sci U S A 2011; 108:3964-9. [PMID: 21368144 DOI: 10.1073/pnas.1100950108] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cytochrome c oxidase (CcO) is the terminal enzyme of the respiratory chain. This redox-driven proton pump catalyzes the four-electron reduction of molecular oxygen to water, one of the most fundamental processes in biology. Elucidation of the intermediate structures in the catalytic cycle is crucial for understanding both the mechanism of oxygen reduction and its coupling to proton pumping. Using CcO from Paracoccus denitrificans, we demonstrate that the artificial F state, classically generated by reaction with an excess of hydrogen peroxide, can be converted into a new P state (in contradiction to the conventional direction of the catalytic cycle) by addition of ammonia at pH 9. We suggest that ammonia coordinates directly to Cu(B) in the binuclear active center in this P state and discuss the chemical structures of both oxoferryl intermediates F and P. Our results are compatible with a superoxide bound to Cu(B) in the F state.
Collapse
|
13
|
Koepke J, Olkhova E, Angerer H, Müller H, Peng G, Michel H. High resolution crystal structure of Paracoccus denitrificans cytochrome c oxidase: new insights into the active site and the proton transfer pathways. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:635-45. [PMID: 19374884 DOI: 10.1016/j.bbabio.2009.04.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 04/03/2009] [Accepted: 04/08/2009] [Indexed: 11/19/2022]
Abstract
The structure of the two-subunit cytochrome c oxidase from Paracoccus denitrificans has been refined using X-ray cryodata to 2.25 A resolution in order to gain further insights into its mechanism of action. The refined structural model shows a number of new features including many additional solvent and detergent molecules. The electron density bridging the heme a(3) iron and Cu(B) of the active site is fitted best by a peroxo-group or a chloride ion. Two waters or OH(-) groups do not fit, one water (or OH(-)) does not provide sufficient electron density. The analysis of crystals of cytochrome c oxidase isolated in the presence of bromide instead of chloride appears to exclude chloride as the bridging ligand. In the D-pathway a hydrogen bonded chain of six water molecules connects Asn131 and Glu278, but the access for protons to this water chain is blocked by Asn113, Asn131 and Asn199. The K-pathway contains two firmly bound water molecules, an additional water chain seems to form its entrance. Above the hemes a cluster of 13 water molecules is observed which potentially form multiple exit pathways for pumped protons. The hydrogen bond pattern excludes that the Cu(B) ligand His326 is present in the imidazolate form.
Collapse
Affiliation(s)
- Juergen Koepke
- Max Planck Institute of Biophysics, Department of Molecular Membrane Biology, Max-von-Laue-Str.3, D-60438 Frankfurt/Main, Germany
| | | | | | | | | | | |
Collapse
|
14
|
Conrath K, Pereira AS, Martins CE, Timóteo CG, Tavares P, Spinelli S, Kinne J, Flaudrops C, Cambillau C, Muyldermans S, Moura I, Moura JJG, Tegoni M, Desmyter A. Camelid nanobodies raised against an integral membrane enzyme, nitric oxide reductase. Protein Sci 2009; 18:619-28. [PMID: 19241371 PMCID: PMC2760367 DOI: 10.1002/pro.69] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 12/18/2008] [Indexed: 12/11/2022]
Abstract
Nitric Oxide Reductase (NOR) is an integral membrane protein performing the reduction of NO to N(2)O. NOR is composed of two subunits: the large one (NorB) is a bundle of 12 transmembrane helices (TMH). It contains a b type heme and a binuclear iron site, which is believed to be the catalytic site, comprising a heme b and a non-hemic iron. The small subunit (NorC) harbors a cytochrome c and is attached to the membrane through a unique TMH. With the aim to perform structural and functional studies of NOR, we have immunized dromedaries with NOR and produced several antibody fragments of the heavy chain (VHHs, also known as nanobodies). These fragments have been used to develop a faster NOR purification procedure, to proceed to crystallization assays and to analyze the electron transfer of electron donors. BIAcore experiments have revealed that up to three VHHs can bind concomitantly to NOR with affinities in the nanomolar range. This is the first example of the use of VHHs with an integral membrane protein. Our results indicate that VHHs are able to recognize with high affinity distinct epitopes on this class of proteins, and can be used as versatile and valuable tool for purification, functional study and crystallization of integral membrane proteins.
Collapse
Affiliation(s)
- Katja Conrath
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit BrusselPleinlaan 2, B-1050 Brussels, Belgium
- Department of Molecular and Cellular InteractionsVIB, Brussels, Belgium
| | - Alice S Pereira
- REQUIMTE, Departamento de Química, Centro de Química Fina e Biotecnologia, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa2829-516 Caparica, Portugal
| | - Carlos E Martins
- REQUIMTE, Departamento de Química, Centro de Química Fina e Biotecnologia, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa2829-516 Caparica, Portugal
| | - Cristina G Timóteo
- REQUIMTE, Departamento de Química, Centro de Química Fina e Biotecnologia, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa2829-516 Caparica, Portugal
| | - Pedro Tavares
- REQUIMTE, Departamento de Química, Centro de Química Fina e Biotecnologia, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa2829-516 Caparica, Portugal
| | - Silvia Spinelli
- Architecture et Fonction des Macromolécules Biologiques, UMR 6098 CNRS and Universités of MarseilleMarseille, France
| | - Joerg Kinne
- Central Veterinary Research LaboratoryDubai, United Arab Emirates
| | - Christophe Flaudrops
- Architecture et Fonction des Macromolécules Biologiques, UMR 6098 CNRS and Universités of MarseilleMarseille, France
| | - Christian Cambillau
- Architecture et Fonction des Macromolécules Biologiques, UMR 6098 CNRS and Universités of MarseilleMarseille, France
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit BrusselPleinlaan 2, B-1050 Brussels, Belgium
- Department of Molecular and Cellular InteractionsVIB, Brussels, Belgium
| | - Isabel Moura
- REQUIMTE, Departamento de Química, Centro de Química Fina e Biotecnologia, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa2829-516 Caparica, Portugal
| | - Jose J G Moura
- REQUIMTE, Departamento de Química, Centro de Química Fina e Biotecnologia, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa2829-516 Caparica, Portugal
| | - Mariella Tegoni
- Architecture et Fonction des Macromolécules Biologiques, UMR 6098 CNRS and Universités of MarseilleMarseille, France
| | - Aline Desmyter
- Architecture et Fonction des Macromolécules Biologiques, UMR 6098 CNRS and Universités of MarseilleMarseille, France
| |
Collapse
|
15
|
Dürr KL, Koepke J, Hellwig P, Müller H, Angerer H, Peng G, Olkhova E, Richter OMH, Ludwig B, Michel H. A D-Pathway Mutation Decouples the Paracoccus denitrificans Cytochrome c Oxidase by Altering the Side-Chain Orientation of a Distant Conserved Glutamate. J Mol Biol 2008; 384:865-77. [DOI: 10.1016/j.jmb.2008.09.074] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 08/28/2008] [Accepted: 09/17/2008] [Indexed: 11/16/2022]
|
16
|
Nauli S, Farr S, Lee YJ, Kim HY, Faham S, Bowie JU. Polymer-driven crystallization. Protein Sci 2007; 16:2542-51. [PMID: 17962407 PMCID: PMC2211692 DOI: 10.1110/ps.073074207] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Revised: 07/31/2007] [Accepted: 08/01/2007] [Indexed: 10/22/2022]
Abstract
Obtaining well-diffracting crystals of macromolecules remains a significant barrier to structure determination. Here we propose and test a new approach to crystallization, in which the crystallization target is fused to a polymerizing protein module, so that polymer formation drives crystallization of the target. We test the approach using a polymerization module called 2TEL, which consists of two tandem sterile alpha motif (SAM) domains from the protein translocation Ets leukemia (TEL). The 2TEL module is engineered to polymerize as the pH is lowered, which allows the subtle modulation of polymerization needed for crystal formation. We show that the 2TEL module can drive the crystallization of 11 soluble proteins, including three that resisted prior crystallization attempts. In addition, the 2TEL module crystallizes in the presence of various detergents, suggesting that it might facilitate membrane protein crystallization. The crystal structures of two fusion proteins show that the TELSAM polymer is responsible for the majority of contacts in the crystal lattice. The results suggest that biological polymers could be designed as crystallization modules.
Collapse
Affiliation(s)
- Sehat Nauli
- UCLA-DOE Institute of Genomics and Proteomics, University of California, Los Angeles 90095-1570, USA
| | | | | | | | | | | |
Collapse
|
17
|
Plewnia G, Schulze K, Hunte C, Tampé R, Koch J. Modulation of the antigenic peptide transporter TAP by recombinant antibodies binding to the last five residues of TAP1. J Mol Biol 2007; 369:95-107. [PMID: 17418234 DOI: 10.1016/j.jmb.2007.02.102] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 02/19/2007] [Accepted: 02/23/2007] [Indexed: 11/30/2022]
Abstract
The transporter associated with antigen processing (TAP) plays a pivotal role in the major histocompatibility complex (MHC) class I mediated immune response against infected or malignantly transformed cells. It belongs to the ATP-binding cassette (ABC) superfamily and consists of TAP1 (ABCB2) and TAP2 (ABCB3), each of which possesses a transmembrane and a nucleotide-binding domain (NBD). Here we describe the generation of recombinant Fv and Fab antibody fragments to human TAP from a hybridoma cell line expressing the TAP1-specific monoclonal antibody mAb148.3. The epitope of the antibody was mapped to the very last five C-terminal amino acid residues of TAP1 on solid-supported peptide arrays. The recombinant antibody fragments were heterologously expressed in Escherichia coli and purified to homogeneity from periplasmic extracts by affinity chromatography. The monoclonal and recombinant antibodies bind with nanomolar affinity to the last five C-terminal amino acid residues of TAP1 as demonstrated by ELISA and surface plasmon resonance. Strikingly, the recombinant antibody fragments confer thermal stability to the heterodimeric TAP complex. At the same time TAP is arrested in a peptide transport incompetent conformation, although ATP and peptide binding to TAP are not affected. Based on our results we suggest that the C terminus of TAP1 modulates TAP function presumably as part of the dimer interface of the NBDs.
Collapse
Affiliation(s)
- Gabriele Plewnia
- Institute of Biochemistry, Biocenter, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 9, D-69438 Frankfurt a. M., Germany
| | | | | | | | | |
Collapse
|
18
|
MacMillan F, Budiman K, Angerer H, Michel H. The role of tryptophan 272 in theParacoccus denitrificanscytochromecoxidase. FEBS Lett 2006; 580:1345-9. [PMID: 16460733 DOI: 10.1016/j.febslet.2006.01.054] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Revised: 01/18/2006] [Accepted: 01/18/2006] [Indexed: 11/22/2022]
Abstract
The mechanism of electron coupled proton transfer in cytochrome c oxidase (CcO) is still poorly understood. The P(M)-intermediate of the catalytic cycle is an oxoferryl state whose generation requires one additional electron, which cannot be provided by the two metal centres. The missing electron has been suggested to be donated to this binuclear site by a tyrosine residue. A tyrosine radical species has been detected in the P(M) and F* intermediates (formed by addition of H2O2) of the Paraccocus denitrificans CcO using electron paramagnetic resonance (EPR) spectroscopy. From the study of conserved variants its origin was determined to be Y167 which is surprising as this residue is not part of the active site. Upon inspection of the active site it becomes evident that W272 could be the actual donor of the missing electron, which can then be replenished from Y167 or from the Y280-H276 cross link in the natural cycle. To address the question, whether such a direct electron transfer pathway to the binuclear centre exists two tryptophan 272 variants in subunit I have been generated. These variants are characterised by their turnover rates as well as using EPR and optical spectroscopy. From these experiments it is concluded, that W272 is an important intermediate in the formation of the radical species appearing in P(M) and F* intermediates produced with hydrogen peroxide. The significance of this finding for the catalytic function of the enzyme is discussed.
Collapse
Affiliation(s)
- Fraser MacMillan
- Institut für Physikalische and Theoretische Chemie, Center for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe-Universität, Frankfurt D-60439, Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
19
|
Zuber D, Krause R, Venturi M, Padan E, Bamberg E, Fendler K. Kinetics of charge translocation in the passive downhill uptake mode of the Na+/H+ antiporter NhaA of Escherichia coli. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2005; 1709:240-50. [PMID: 16139785 DOI: 10.1016/j.bbabio.2005.07.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Revised: 07/26/2005] [Accepted: 07/27/2005] [Indexed: 11/24/2022]
Abstract
The Na+/H+ antiporter NhaA is the main Na+ extrusion system in E. coli. Using direct current measurements combined with a solid supported membrane (SSM), we obtained electrical data of the function of NhaA purified and reconstituted in liposomes. These measurements demonstrate NhaA's electrogenicity, its specificity for Li+ and Na+ and its pronounced pH dependence in the range pH 6.5-8.5. The mutant G338S, in contrast, presents a pH independent profile, as reported previously. A complete right-side-out orientation of the NhaA antiporter within the proteoliposomal membrane was determined using a NhaA-specific antibody based ELISA assay. This allowed for the first time the investigation of NhaA in the passive downhill uptake mode corresponding to the transport of Na+ from the periplasmic to the cytoplasmic side of the membrane. In this mode, the transporter has kinetic properties differing significantly from those of the previously investigated efflux mode. The apparent Km values were 11 mM for Na+ and 7.3 mM for Li+ at basic pH and 180 mM for Na+ and 50 mM for Li+ at neutral pH. The data demonstrate that in the passive downhill uptake mode pH regulation of the carrier affects both apparent Km as well as turnover (Vmax).
Collapse
Affiliation(s)
- D Zuber
- Max Planck Institut für Biophysik, Max von Laue Strasse 3, D-60438 Frankfurt/Main, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Richter OMH, Dürr KL, Kannt A, Ludwig B, Scandurra FM, Giuffrè A, Sarti P, Hellwig P. Probing the access of protons to the K pathway in the Paracoccus denitrificans cytochrome c oxidase. FEBS J 2005; 272:404-12. [PMID: 15654878 DOI: 10.1111/j.1742-4658.2004.04480.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In recent studies on heme-copper oxidases a particular glutamate residue in subunit II has been suggested to constitute the entry point of the so-called K pathway. In contrast, mutations of this residue (E78(II)) in the Paracoccus denitrificans cytochrome c oxidase do not affect its catalytic activity at all (E78(II)Q) or reduce it to about 50% (E78(II)A); in the latter case, the mutation causes no drastic decrease in heme a(3) reduction kinetics under anaerobic conditions, when compared to typical K pathway mutants. Moreover, both mutant enzymes retain full proton-pumping competence. While oxidized-minus-reduced Fourier-transform infrared difference spectroscopy demonstrates that E78(II) is indeed addressed by the redox state of the enzyme, absence of variations in the spectral range characteristic for protonated aspartic and glutamic acids at approximately 1760 to 1710 cm(-1) excludes the protonation of E78(II) in the course of the redox reaction in the studied pH range, although shifts of vibrational modes at 1570 and 1400 cm(-1) reflect the reorganization of its deprotonated side chain at pH values greater than 4.8. We therefore conclude that protons do not enter the K channel via E78(II) in the Paracoccus enzyme.
Collapse
Affiliation(s)
- Oliver-M H Richter
- Institut für Biochemie, Abteilung Molekulare Genetik, Johann Wolfgang Goethe-Universität, Frankfurt-am-Main, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Kashino Y. Separation methods in the analysis of protein membrane complexes. J Chromatogr B Analyt Technol Biomed Life Sci 2004; 797:191-216. [PMID: 14630150 DOI: 10.1016/s1570-0232(03)00428-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The separation of membrane protein complexes can be divided into two categories. One category, which is operated on a relatively large scale, aims to purify the membrane protein complex from membrane fractions while retaining its native form, mainly to characterize its nature. The other category aims to analyze the constituents of the membrane protein complex, usually on a small scale. Both of these face the difficulty of isolating the membrane protein complex without interference originating from the hydrophobic nature of membrane proteins or from the close association with membrane lipids. To overcome this difficulty, many methods have been employed. Crystallized membrane protein complexes are the most successful example of the former category. In these purification methods, special efforts are made in the steps prior to the column chromatography to enrich the target membrane protein complexes. Although there are specific aspects for each complex, the most popular method for isolating these membrane protein complexes is anion-exchange column chromatography, especially using weak anion-exchange columns. Another remarkable trend is metal affinity column chromatography, which purifies the membrane protein complex as an intact complex in one step. Such protein complexes contain subunit proteins which are genetically engineered so as to include multiple-histidine tags at carboxyl- or amino-termini. The key to these successes for multi-subunit complex isolation is the idea of keeping the expression at its physiological level, rather than overexpression. On the other hand, affinity purification using the Fv fragment, in which a Strep tag is genetically introduced, is ideal because this method does not introduce any change to the target protein. These purification methods supported by affinity interaction can be applied to minor membrane protein complexes in the membrane system. Isoelectric focusing (IEF) and blue native (BN) electrophoresis have also been employed to prepare membrane protein complexes. Generally, a combination of two or more chromatographic and/or electrophoretic methods is conducted to separate membrane protein complexes. IEF or BN electrophoresis followed by 2nd dimension electrophoresis serve as useful tools for analytical demand. However, some problems still exist in the 2D electrophoresis using IEF. To resolve such problems, many attempts have been made, e.g. introduction of new chaotropes, surfactants, reductants or supporting matrices. This review will focus in particular on two topics: the preparative methods that achieved purification of membrane protein complexes in the native (intact) form, and the analytical methods oriented to resolve the membrane proteins. The characteristics of these purification and analytical methods will be discussed along with plausible future developments taking into account the nature of membrane protein complexes.
Collapse
Affiliation(s)
- Yasuhiro Kashino
- Faculty of Science, Department of Life Science, Himeji Institute of Technology, Harima Science Garden City, Hyogo 678-1297, Japan.
| |
Collapse
|
22
|
Venturi M, Hunte C. Monoclonal antibodies for the structural analysis of the Na+/H+ antiporter NhaA from Escherichia coli. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1610:46-50. [PMID: 12586378 DOI: 10.1016/s0005-2736(02)00713-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Since their advent some 25 years ago, monoclonal antibodies have developed into powerful tools for structural and functional analysis of their cognate antigens. Together with the respective antigen binding fragments, antibodies offer exclusive capacities in detection, characterization, purification and functional assays for every given ligand. Antibody-fragment mediated crystallization represents a major advance in determining the three-dimensional structure of membrane-bound protein complexes. In this review, we focus on the methods used to generate monoclonal antibodies against the NhaA antiporter from Escherichia coli as a paradigm of secondary transporters. We describe examples on how antibodies are helpful in understanding structure and function relationships for this important class of integral membrane proteins. The generated conformation-specific antibody fragments are highly valuable reagents for co-crystallization attempts and structure determination of the antiporter.
Collapse
Affiliation(s)
- Miro Venturi
- Structural Biology Section, Vaccine Research Center/National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
23
|
Engel CK, Chen L, Privé GG. Insertion of carrier proteins into hydrophilic loops of the Escherichia coli lactose permease. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1564:38-46. [PMID: 12100994 DOI: 10.1016/s0005-2736(02)00398-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We describe the design and characterization of a set of fusion proteins of the Escherichia coli lactose (lac) permease in which a set of five different soluble "carrier" proteins (cytochrome(b562), flavodoxin, T4 lysozyme, beta-lactamase and 70 kDa heat shock ATPase domain) were systematically inserted into selected loop positions of the transporter. The design goal was to increase the exposed hydrophilic surface area of the permease, while minimizing the internal flexibility of the resulting fusion proteins in order to improve the crystallization properties of the membrane protein. Fusion proteins with insertions into the central hydrophilic loop of the lac permease were active in transport lactose, although only the fusion proteins with E. coli cytochrome(b562), E. coli flavodoxin or T4 lysozyme were expressed at near wild-type lac permease levels. Eight other loop positions were tested with these three carriers, leading to the identification of additional fusion proteins that were active and well-expressed. By combining the results from the single carrier insertions, we have expressed functional "double fusion" proteins containing cytochrome(b562) domains inserted in two different loop positions.
Collapse
Affiliation(s)
- Christian K Engel
- Division of Molecular and Structural Biology, Ontario Cancer Institute, Toronto, Canada
| | | | | |
Collapse
|
24
|
Drosou V, Malatesta F, Ludwig B. Mutations in the docking site for cytochrome c on the Paracoccus heme aa3 oxidase. Electron entry and kinetic phases of the reaction. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:2980-8. [PMID: 12071962 DOI: 10.1046/j.1432-1033.2002.02979.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Introducing site-directed mutations in surface-exposed residues of subunit II of the heme aa3 cytochrome c oxidase of Paracoccus denitrificans, we analyze the kinetic parameters of electron transfer from reduced horse heart cytochrome c. Specifically we address the following issues: (a) which residues on oxidase contribute to the docking site for cytochrome c, (b) is an aromatic side chain required for electron entry from cytochrome c, and (c) what is the molecular basis for the previously observed biphasic reaction kinetics. From our data we conclude that tryptophan 121 on subunit II is the sole entry point for electrons on their way to the CuA center and that its precise spatial arrangement, but not its aromatic nature, is a prerequisite for efficient electron transfer. With different reaction partners and experimental conditions, biphasicity can always be induced and is critically dependent on the ionic strength during the reaction. For an alternative explanation to account for this phenomenon, we find no evidence for a second cytochrome c binding site on oxidase.
Collapse
Affiliation(s)
- Viktoria Drosou
- Molecular Genetics, Institute of Biochemistry, Johann-Wolfgang-Goethe Universität, Frankfurt, Germany
| | | | | |
Collapse
|
25
|
Blank K, Lindner P, Diefenbach B, Plückthun A. Self-immobilizing recombinant antibody fragments for immunoaffinity chromatography: generic, parallel, and scalable protein purification. Protein Expr Purif 2002; 24:313-22. [PMID: 11858727 DOI: 10.1006/prep.2001.1575] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We present the directed immobilization of recombinant antibody fragments as ligands for general immunoaffinity chromatography methods. It is based on fusion proteins of scFv fragments with several chitin-binding domains which can be immobilized directly from a crude bacterial lysate on inexpensive chitin beads for the purification of proteins without any gradient or detector. It has been used with a positive pressure manifold, allowing the parallel processing of 24 different samples on a milligram scale, as convenient as plasmid isolation. The method is demonstrated with several anti-protein antibodies. In addition, methods are presented of using an anti-His tag antibody either alone or directly coupled to IMAC to obtain very pure protein. As those methods are scalable, they should prove very useful in the parallel purification of natural and recombinant proteins on small scales (for proteomics), medium scales (for crystallography and NMR), and very large scales (for therapeutic proteins).
Collapse
Affiliation(s)
- Kerstin Blank
- Biochemisches Institut, Universität Zürich, Switzerland
| | | | | | | |
Collapse
|
26
|
Venturi M, Seifert C, Hunte C. High level production of functional antibody Fab fragments in an oxidizing bacterial cytoplasm. J Mol Biol 2002; 315:1-8. [PMID: 11771962 DOI: 10.1006/jmbi.2001.5221] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The antigen-binding fragments (Fab) of antibodies are powerful tools in clinical therapy, molecular diagnostics and basic research. However, their principal applications require pure recombinant molecules in large amounts, which are challenging to obtain. Severe limitations in yield, folding and functionality are commonly encountered in bacterial production of Fab fragments. Secretion into the oxidizing periplasm generally results in low yield, whereas expression in the reducing cytoplasmic environment produces unfolded or non-functional protein. We hypothesized that an impaired reducing environment of the cytoplasm would permit correctly folded, functional cytoplasmic expression of Fabs with high yield. We used the Escherichia coli strain FA113, which has no activity of both thioredoxin and glutathione reductase, and thus has an oxidizing cytoplasmic environment. With the newly constructed vector pFAB1 we tested the cytoplasmic expression of two Fab fragments, which recognize the integral membrane protein NhaA, a bacterial Na(+)/H(+) antiporter. These antibodies differ in terms of DNA sequence and stability. Both antibody fragments were produced to very high yields (10-30 mg/l from bacterial cultures at an A(600 nm)=1.2-1.3). This is a factor 50-250 times higher than any other reported over-expression strategy for Fab fragments and currently represents the highest production rate ever been reported for antibody Fab fragments in bacteria grown to similar cell densities. The fragments are fully functional and can be efficiently purified by His-tag chromatography. Expression of active Fab fragments in the bacterial cytoplasm unlocks the possibility of using antibody specific targeting in an intracellular environment. Such a capacity opens new perspectives for investigating metabolic and regulatory pathways in vivo and also provides a powerful selection system for functional genomics.
Collapse
Affiliation(s)
- Miro Venturi
- Max-Planck-Institut für Biophysik, Abteilung für Molekulare Membranbiologie, Heinrich-Hoffmann-Str.7, Frankfurt am Main, D-60528, Germany
| | | | | |
Collapse
|
27
|
Hellwig P, Rost B, Mäntele W. Redox dependent conformational changes in the mixed valence form of the cytochrome c oxidase from p. The reorganization of glutamic acid 278 is coupled to the electron transfer from/to heme a and the binuclear center. denitrificans. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2001; 57A:1123-1131. [PMID: 11374571 DOI: 10.1016/s1386-1425(00)00472-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
In this work we present the separation of FTIR difference signals induced by electron transfer to/from the redox centers of the cytochrome c oxidase from P. denitrificans and compare electrochemically induced FTIR difference spectra with those induced by CO photolysis. FTIR difference spectra of rebinding of CO to the half reduced (mixed valence) form of the cytochrome c oxidase after photolysis reflect the conformational changes induced by the rebinding of CO and by electron transfer reactions from heme a3 to heme a and further on to CUA. During this process, heme a3 (and CUB) are oxidized, whereas heme a and CuA are reduced. By subtracting these difference spectra from an electrochemically induced FTIR difference spectrum, where all four cofactors are reduced, the contributions for heme a3 (and CuB) could be separated. Correspondingly, the spectral contributions of heme a and CuA have been separated. The comparison of these spectra with the spectra calculated for the hemes on the basis of their redox dependent changes previously published in Hellwig et al., (Biochemistry 38, (1999) 1685-1694) show a high degree of similarity, except for additional signals coupled to the reorganization of the binuclear center upon CO rebinding. The separated spectra clearly show that the signals attributed to Glu278, an amino acid discussed to be crucial for proton pumping, is coupled to electron transfer to/from heme a and the binuclear heme a3-CuB center.
Collapse
Affiliation(s)
- P Hellwig
- Institut für Biophysik, Johann-Wolfgang-Goethe-Universität, Frankfurt, Germany.
| | | | | |
Collapse
|
28
|
Skerra A, Schmidt TG. Use of the Strep-Tag and streptavidin for detection and purification of recombinant proteins. Methods Enzymol 2001; 326:271-304. [PMID: 11036648 DOI: 10.1016/s0076-6879(00)26060-6] [Citation(s) in RCA: 179] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- A Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Freising-Weihenstephan, Germany
| | | |
Collapse
|
29
|
Hunte C, Koepke J, Lange C, Rossmanith T, Michel H. Structure at 2.3 A resolution of the cytochrome bc(1) complex from the yeast Saccharomyces cerevisiae co-crystallized with an antibody Fv fragment. Structure 2000; 8:669-84. [PMID: 10873857 DOI: 10.1016/s0969-2126(00)00152-0] [Citation(s) in RCA: 447] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND The cytochrome bc(1) complex is part of the energy conversion machinery of the respiratory and photosynthetic electron transfer chains. This integral membrane protein complex catalyzes electron transfer from ubiquinol to cytochrome c. It couples the electron transfer to the electrogenic translocation of protons across the membrane via a so-called Q cycle mechanism. RESULTS The cytochrome bc(1) complex from the yeast Saccharomyces cerevisiae was crystallized together with a bound antibody Fv fragment. The structure was determined at 2.3 A resolution using multiple isomorphous replacement, and refined to a crystallographic R factor of 22.2% (R(free) = 25.4%). The complex is present as a homodimer. Each 'monomer' of the refined model includes 2178 amino acid residues of subunits COR1, QCR2, COB, CYT1, RIP1, QCR6, QCR7, QCR8 and QCR9 of the cytochrome bc(1) complex and of the polypeptides V(H) and V(L) of the Fv fragment, the cofactors heme b(H), heme b(L), heme c(1), the [2Fe-2S] cluster and 346 water molecules. The Fv fragment binds to the extrinsic domain of the [2Fe-2S] Rieske protein and is essential for formation of the crystal lattice. CONCLUSIONS The approach to crystallize membrane proteins as complexes with specific antibody fragments appears to be of general importance. The structure of the yeast cytochrome bc(1) complex reveals in detail the binding sites of the natural substrate coenzyme Q6 and the inhibitor stigmatellin. Buried water molecules close to the binding sites suggest possible pathways for proton uptake and release. A comparison with other cytochrome bc(1) complexes shows features that are specific to yeast.
Collapse
Affiliation(s)
- C Hunte
- Max-Planck-Institut für Biophysik, Abt. Molekulare Membranbiologie, Frankfurt, 60528, Germany.
| | | | | | | | | |
Collapse
|
30
|
Ruitenberg M, Kannt A, Bamberg E, Ludwig B, Michel H, Fendler K. Single-electron reduction of the oxidized state is coupled to proton uptake via the K pathway in Paracoccus denitrificans cytochrome c oxidase. Proc Natl Acad Sci U S A 2000; 97:4632-6. [PMID: 10781069 PMCID: PMC18284 DOI: 10.1073/pnas.080079097] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2000] [Indexed: 11/18/2022] Open
Abstract
The reductive part of the catalytic cycle of cytochrome c oxidase from Paracoccus denitrificans was examined by using time-resolved potential measurements on black lipid membranes. Proteoliposomes were adsorbed to the black lipid membranes and Ru(II)(2, 2'-bipyridyl)(3)(2+) was used as photoreductant to measure flash-induced membrane potential generation. Single-electron reduction of the oxidized wild-type cytochrome c oxidase reveals two phases of membrane potential generation (tau(1) approximately 20 micros and tau(2) approximately 175 micros) at pH 7.4. The fast phase is not sensitive to cyanide and is assigned to electron transfer from Cu(A) to heme a. The slower phase is inhibited completely by cyanide and shows a kinetic deuterium isotope effect by a factor of 2-3. Although two enzyme variants mutated in the so-called D pathway of proton transfer (D124N and E278Q) show the same time constants and relative amplitudes as the wild-type enzyme, in the K pathway variant K354M, tau(2) is increased to 900 micros. This result suggests uptake of a proton through the K pathway during the transition from the oxidized to the one-electron reduced state. After the second laser flash under anaerobic conditions, a third electrogenic phase with a time constant of approximately 1 ms appears. The amplitude of this phase grows with increasing flash number. We explain this growth by injection of a second electron into the single-electron reduced enzyme. On multiple flashes, both D pathway mutants behave differently compared with the wild type and two additional slow phases of tau(3) approximately 2 ms and tau(4) approximately 15 ms are observed. These results suggest that the D pathway is involved in proton transfer coupled to the uptake of the second electron.
Collapse
Affiliation(s)
- M Ruitenberg
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry, Kennedyallee 70, 60596 Frankfurt/Main, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Huber O, Huber-Wunderlich M. Recombinant Proteins. ACTA ACUST UNITED AC 2000. [DOI: 10.1016/s0301-4770(08)60541-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
32
|
Skerra A, Schmidt TG. Applications of a peptide ligand for streptavidin: the Strep-tag. BIOMOLECULAR ENGINEERING 1999; 16:79-86. [PMID: 10796988 DOI: 10.1016/s1050-3862(99)00033-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The Strep-tag constitutes a nine amino acid-peptide that binds specifically to streptavidin and occupies the same pocket where biotin is normally complexed. Since the Strep-tag participates in a reversible interaction it can be applied for the efficient purification of corresponding fusion proteins on affinity columns with immobilized streptavidin. Elution of the bound recombinant protein can be effected under mild buffer conditions by competition with biotin or a suitable derivative. In addition, Strep-tag fusion proteins can be easily detected in immunochemical assays, like Western blots or ELISAs, by means of commercially available streptavidin-enzyme conjugates. The Strep-tag/streptavidin system has been systematically optimized over the past years, including the engineering of streptavidin itself. Structural insight into the molecular mimicry between the peptide and biotin was furthermore gained from X-ray crystallographic analysis. As a result the system provides a reliable and versatile tool in recombinant protein chemistry. Exemplary applications of the Strep-tag are discussed in this review.
Collapse
Affiliation(s)
- A Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Freising-Weihenstephan, Germany
| | | |
Collapse
|
33
|
Nicoletti F, Witt H, Ludwig B, Brunori M, Malatesta F. Paracoccus denitrificans cytochrome c oxidase: a kinetic study on the two- and four-subunit complexes. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1365:393-403. [PMID: 9757081 DOI: 10.1016/s0005-2728(98)00092-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cytochrome c oxidase from Paracoccus denitrificans has been purified in two different forms differing in polypeptide composition. An enzyme containing polypeptides I-IV is obtained when the purification procedure is performed in beta-d-dodecylmaltoside. If, however, Triton X-100 is used to purify the enzyme under otherwise identical conditions, an enzyme is obtained containing only subunits I-II. The two enzymes are undistinguishable by optical spectroscopy but show significant differences in the transient and steady-state time regimes, as studied by stopped-flow spectroscopy. The observed differences, however, are not due to removal of subunits III and IV, but rather to a specific effect of Triton X-100 which appears to affect cytochrome c binding. From these results it is not expected that subunits III and IV play any significant role in cytochrome c binding and, possibly, in the subsequent electron transfer processes. The results also suggest that both electrostatic and hydrophobic interactions may be important in the initial electron transfer process from cytochrome c.
Collapse
Affiliation(s)
- F Nicoletti
- Department of Biochemical Sciences and CNR Centre of Molecular Biology, University of Rome 'La Sapienza', Piazzale A. Moro, 5, 00185 Rome, Italy
| | | | | | | | | |
Collapse
|
34
|
Abstract
We have previously described the isolation and in vitro binding properties of eight anti-DNA monoclonal antibodies (MAbs) from an MRL-lpr mouse. In light of recent reports that have indicated it is possible to isolate multiple MAbs from a single hybridoma, our pathogenic hybridoma, 11F8, was examined for evidence of similar phenomena. Chromosome counting suggested that 11F8 cells are unusual and might indeed be expressing multiple heavy and/or light chains. PCR, cloning, and sequencing of immunoglobulin heavy and light chains indicate that 11F8 displays expression of both gamma 2a and gamma 3 heavy chains at the DNA level. Flow cytometry and amino acid sequencing reveals that expression of multiple isotypes also occurs at the protein level but only a single heavy- and light-chain sequence is able to bind DNA. Based on these results, we conclude that 11F8 is an unusual hybridoma that secretes two distinct heavy and at least one light chain from a single cell, and may represent a trioma, a stable three-cell fusion.
Collapse
Affiliation(s)
- N B Blatt
- University of Michigan, Department of Chemistry, Ann Arbor 48109-1055, USA
| | | | | |
Collapse
|
35
|
Pfitzner U, Odenwald A, Ostermann T, Weingard L, Ludwig B, Richter OM. Cytochrome c oxidase (heme aa3) from Paracoccus denitrificans: analysis of mutations in putative proton channels of subunit I. J Bioenerg Biomembr 1998; 30:89-97. [PMID: 9623810 DOI: 10.1023/a:1020515713103] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One of the challenging features of energy-transducing terminal oxidases, like the aa3 cytochrome c oxidase of Paracoccus denitrificans, is the translocation of protons across the cytoplasmic membrane, which is coupled to the transfer of electrons to oxygen. As a prerequisite for a more advanced examination of the enzymatic properties, several amino acid residues, selected on the basis of recent three-dimensional structure determinations, were exchanged in subunit I of the Paracoccus enzyme by site-directed mutagenesis. The properties of the mutated oxidases were analyzed by different methods to elucidate whether they are involved in the coupled and coordinated transfer of protons via two different pathways either to the site of oxygen reduction or through the enzyme from the cytoplasm to the periplasmic side.
Collapse
Affiliation(s)
- U Pfitzner
- Molecular Genetics, Institute of Biochemistry, Biozentrum, Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
36
|
Dennison C, Canters G, Vries S, Vijgenboom E, Spanning R. The Methylamine Dehydrogenase Electron Transfer Chain. ADVANCES IN INORGANIC CHEMISTRY 1998. [DOI: 10.1016/s0898-8838(08)60029-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
37
|
Abstract
Five new membrane protein structures have been determined since 1995 using X-ray crystallography: bacterial light-harvesting complex; bacterial and mitochondrial cytochrome c oxidases; mitochondrial bc1 complex; and alpha-hemolysin. These successes are partly based on advances in the crystallization procedures for integral membrane proteins. Variation of the size of the detergent micelle and/or increasing the size of the polar surface of the membrane protein is the most important route to well-ordered membrane protein crystals. The use of bicontinuous lipidic cubic phases also appears to be promising.
Collapse
Affiliation(s)
- C Ostermeier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520-8114, USA.
| | | |
Collapse
|
38
|
Nilsson J, Ståhl S, Lundeberg J, Uhlén M, Nygren PA. Affinity fusion strategies for detection, purification, and immobilization of recombinant proteins. Protein Expr Purif 1997; 11:1-16. [PMID: 9325133 DOI: 10.1006/prep.1997.0767] [Citation(s) in RCA: 232] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- J Nilsson
- Department of Biochemistry and Biotechnology, Royal Institute of Technology (KTH), Stockholm, Sweden
| | | | | | | | | |
Collapse
|
39
|
Ostermeier C, Harrenga A, Ermler U, Michel H. Structure at 2.7 A resolution of the Paracoccus denitrificans two-subunit cytochrome c oxidase complexed with an antibody FV fragment. Proc Natl Acad Sci U S A 1997; 94:10547-53. [PMID: 9380672 PMCID: PMC23397 DOI: 10.1073/pnas.94.20.10547] [Citation(s) in RCA: 600] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The aa3 type cytochrome c oxidase consisting of the core subunits I and II only was isolated from the soil bacterium Paracoccus denitrificans and crystallized as complex with a monoclonal antibody Fv fragment. Crystals could be grown in the presence of a number of different nonionic detergents. However, only undecyl-beta-D-maltoside and cyclohexyl-hexyl-beta-D-maltoside yielded well-ordered crystals suitable for high resolution x-ray crystallographic studies. The crystals belong to space group P212121 and diffract x-rays to at least 2.5 A (1 A = 0.1 nm) resolution using synchrotron radiation. The structure was determined to a resolution of 2.7 A using molecular replacement and refined to a crystallographic R-factor of 20.5% (Rfree = 25.9%). The refined model includes subunits I and II and the 2 chains of the Fv fragment, 2 heme A molecules, 3 copper atoms, and 1 Mg/Mn atom, a new metal (Ca) binding site, 52 tentatively identified water molecules, and 9 detergent molecules. Only four of the water molecules are located in the cytoplasmic half of cytochrome c oxidase. Most of them are near the interface of subunits I and II. Several waters form a hydrogen-bonded cluster, including the heme propionates and the Mg/Mn binding site. The Fv fragment binds to the periplasmic polar domain of subunit II and is critically involved in the formation of the crystal lattice. The crystallization procedure is well reproducible and will allow for the analysis of the structures of mechanistically interesting mutant cytochrome c oxidases.
Collapse
Affiliation(s)
- C Ostermeier
- Max-Planck-Institut für Biophysik, Abteilung für Molekulare Membranbiologie, Heinrich-Hoffmann-Strasse 7, D-60528 Frankfurt, Germany
| | | | | | | |
Collapse
|
40
|
|
41
|
Tucker J, Grisshammer R. Purification of a rat neurotensin receptor expressed in Escherichia coli. Biochem J 1996; 317 ( Pt 3):891-9. [PMID: 8760379 PMCID: PMC1217569 DOI: 10.1042/bj3170891] [Citation(s) in RCA: 165] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A truncated rat neurotensin receptor (NTR), expressed in Escherichia coli with the maltose-binding protein fused to its N-terminus and the 13 amino acid Bio tag fused to its C-terminus, was purified to apparent homogeneity in two steps by use of the monomeric avidin system followed by a novel neurotensin column. This purification protocol was developed by engineering a variety of affinity tags on to the C-terminus of NTR. Surprisingly, expression levels varied considerably depending on the C-terminal tag used. Functional expression of NTR was highest (800 receptors/cell) when thioredoxin was placed between the receptor C-terminus and the tag, indicating a stabilizing effect of the thioredoxin moiety. Several affinity chromatography methods were tested for purification. NTR with the in vivo-biotinylated Bio tag was purified with the highest efficiency compared with NTR with the Strep tag or a hexa-histidine tail. Co-expression of biotin ligase improved considerably the in vivo biotinylation of the Bio tag and, therefore, the overall purification yield. Proteolysis of the NTR fusion protein was prevented by removing a protease-sensitive site discovered at the N-terminus of NTR. The ligand binding properties of the purified receptor were similar to those of the membrane-bound protein and the native receptor. The scale-up of this purification scheme, to provide sufficient protein for biophysical studies, is in progress.
Collapse
Affiliation(s)
- J Tucker
- Centre for Protein Engineering/MRC Centre, Cambridge, U.K
| | | |
Collapse
|
42
|
Ostermeier C, Michel H. Improved cloning of antibody variable regions from hybridomas by an antisense-directed RNase H digestion of the P3-X63-Ag8.653 derived pseudogene mRNA. Nucleic Acids Res 1996; 24:1979-80. [PMID: 8657583 PMCID: PMC145887 DOI: 10.1093/nar/24.10.1979] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- C Ostermeier
- Max-Planck-Institut für Biophysik, Frankfurt, Germany
| | | |
Collapse
|
43
|
Zickermann V, Verkhovsky M, Morgan J, Wikström M, Anemüller S, Bill E, Steffens GC, Ludwig B. Perturbation of the CuA site in cytochrome-c oxidase of Paracoccus denitrificans by replacement of Met227 with isoleucine. EUROPEAN JOURNAL OF BIOCHEMISTRY 1995; 234:686-93. [PMID: 8536720 DOI: 10.1111/j.1432-1033.1995.686_b.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Subunit II of cytochrome-c oxidase contains a redox centre, CuA, with unusual spectroscopic properties; this site consists of two copper atoms and acts as the entry point for electrons from cytochrome c. We have constructed a site-directed mutant of cytochrome-c oxidase from Paracoccus denitrificans in which the CuA site has been disturbed by replacement of Met227 with isoleucine. The purified, fully assembled enzyme complex has been investigated with various techniques including metal analysis, EPR and visible spectroscopies, steady-state and fast kinetics. The stoichiometry of the metals in the enzyme remains unchanged but a clear perturbation of the CuA site can be observed in the EPR and near-infrared optical spectra. It is concluded that in the mutant CuA is still binuclear but that the two nuclei are no longer equivalent, converting the delocalized [Cu(1.5)....Cu(1.5)] centre of the wild type into a localized [Cu(I)....Cu(II)] system. Changes in the overall kinetics of the mutant are correlated with a diminished electron transfer rate between CuA and heme alpha.
Collapse
Affiliation(s)
- V Zickermann
- Institute of Biochemistry/Molecular Genetics, University of Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Warne A, Wang DN, Saraste M. Purification and two-dimensional crystallization of bacterial cytochrome oxidases. EUROPEAN JOURNAL OF BIOCHEMISTRY 1995; 234:443-51. [PMID: 8536687 DOI: 10.1111/j.1432-1033.1995.443_b.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A novel strategy which employes chromatography on an immobilized metal ion has been developed for the purification of bacterial cytochrome c and quinol oxidases. Many bacterial oxidase complexes appear to have a natural affinity to bind to the chelated copper ion. A combination of three different chromatographic principles (anion exchange, metal-affinity and gel filtration) makes an effective tool chest for the preparation of homogeneous and protein-chemically pure bacterial oxidases. These preparations have been used for two-dimensional crystallization. Until now, crystals have been obtained using the Paracococcus denitrificans and Rhodobacter sphaeroides cytochrome aa3 and the Escherichia coli cytochrome bo. The crystals diffract to approximately 2.5 nm in negative stain and have potential for further structural studies.
Collapse
Affiliation(s)
- A Warne
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | |
Collapse
|
45
|
Tsiotis G, Haase W, Engel A, Michel H. Isolation and Structural Characterization of Trimeric Cyanobacterial Photosystem I Complex with the Help of Recombinant Antibody Fragments. ACTA ACUST UNITED AC 1995. [DOI: 10.1111/j.1432-1033.1995.0823d.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
46
|
Kleymann G, Iwata S, Wiesmüller KH, Ludwig B, Haase W, Michel H. Immunoelectron microscopy and epitope mapping with monoclonal antibodies suggest the existence of an additional N-terminal transmembrane helix in the cytochrome b subunit of bacterial ubiquinol:cytochrome-c oxidoreductases. EUROPEAN JOURNAL OF BIOCHEMISTRY 1995; 230:359-63. [PMID: 7601123 DOI: 10.1111/j.1432-1033.1995.tb20571.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The topology of the ubiquinol:cytochrome-c oxidoreductase (cytochrome bc1 complex) from Paracoccus denitrificans was investigated by immunoelectron microscopy with sequence-specific murine monoclonal antibodies. Epitope mapping with synthetic peptides and enzymic proteolytic cleavage of the cytochrome bc1 complex were employed to localize precisely the respective antibody epitopes on the subunits of this membrane protein complex. Localization of defined epitopes on the cytochrome bc1 complex by immunoelectron microscopy clearly demonstrates that the N-terminus of the cytochrome b subunit is exposed to the periplasmic space. This finding is in agreement with a nine-transmembrane-helices topology model (I-IX) as predicted before for cytochrome b. However, due to other published evidence we favour the existence of an additional transmembrane helix (helix 0) complementing a more recently published eight-helices model (A-C,cd, D-H), at least for prokaryotes.
Collapse
Affiliation(s)
- G Kleymann
- Max-Planck-Institut für Biophysik, Abteilung Molekulare Membranbiologie, Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|